Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Life Sci Alliance ; 4(7)2021 07.
Article En | MEDLINE | ID: mdl-34103390

Haploinsufficiency of progranulin (PGRN) is a leading cause of frontotemporal lobar degeneration (FTLD). PGRN polymorphisms are associated with Alzheimer's disease. PGRN is highly expressed in the microglia near Aß plaques and influences plaque dynamics and microglial activation. However, the detailed mechanisms remain elusive. Here we report that PGRN deficiency reduces human APP and Aß levels in the young male but not female mice. PGRN-deficient microglia exhibit increased expression of markers associated with microglial activation, including CD68, galectin-3, TREM2, and GPNMB, specifically near Aß plaques. In addition, PGRN loss leads to up-regulation of lysosome proteins and an increase in the nuclear localization of TFE3, a transcription factor involved in lysosome biogenesis. Cultured PGRN-deficient microglia show enhanced nuclear translocation of TFE3 and inflammation in response to Aß fibril treatment. Taken together, our data revealed a sex- and age-dependent effect of PGRN on APP metabolism and a role of PGRN in regulating lysosomal activities and inflammation in plaque-associated microglia.


Frontotemporal Lobar Degeneration/metabolism , Plaque, Amyloid/metabolism , Progranulins/metabolism , Age Factors , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/physiology , Amyloid beta-Protein Precursor/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Brain/metabolism , Disease Models, Animal , Female , Frontotemporal Lobar Degeneration/physiopathology , Intercellular Signaling Peptides and Proteins/metabolism , Lysosomes/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , Plaque, Amyloid/physiopathology , Progranulins/physiology , Proteins , Receptors, Immunologic/metabolism , Sex Factors
2.
Cancer Res ; 81(4): 910-922, 2021 02 15.
Article En | MEDLINE | ID: mdl-33323378

Esophageal squamous cell carcinoma (ESCC) is one of the most common and deadly diseases. In our previous comprehensive genomics study, we found that family with sequence similarity 135 member B (FAM135B) was a novel cancer-related gene, yet its biological functions and molecular mechanisms remain unclear. In this study, we demonstrate that the protein levels of FAM135B are significantly higher in ESCC tissues than in precancerous tissues, and high expression of FAM135B correlates with poorer clinical prognosis. Ectopic expression of FAM135B promoted ESCC cell proliferation in vitro and in vivo, likely through its direct interaction with growth factor GRN, thus forming a feedforward loop with AKT/mTOR signaling. Patients with ESCC with overexpression of both FAM135B and GRN had worse prognosis; multivariate Cox model analysis indicated that high expression of both FAM135B and GRN was an independent prognostic factor for patients with ESCC. FAM135B transgenic mice bore heavier tumor burden than wild-type mice and survived a relatively shorter lifespan after 4-nitroquinoline 1-oxide treatment. In addition, serum level of GRN in transgenic mice was higher than in wild-type mice, suggesting that serum GRN levels might provide diagnostic discrimination for patients with ESCC. These findings suggest that the interaction between FAM135B and GRN plays critical roles in the regulation of ESCC progression and both FAM135B and GRN might be potential therapeutic targets and prognostic factors in ESCC. SIGNIFICANCE: These findings investigate the mechanisms of FAM135B in promoting ESCC progression and suggest new potential prognostic biomarkers and therapeutic targets in patients with ESCC.


Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Progranulins/physiology , Animals , Autocrine Communication/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Disease Progression , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/mortality , Female , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Prognosis , Progranulins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism
3.
Mol Cell Neurosci ; 109: 103553, 2020 12.
Article En | MEDLINE | ID: mdl-32956830

Frontotemporal dementia (FTD) describes a group of clinically heterogeneous conditions that frequently affect people under the age of 65 (Le Ber et al., 2013). There are multiple genetic causes of FTD, including coding or splice-site mutations in MAPT, GRN mutations that lead to haploinsufficiency of progranulin protein, and a hexanucleotide GGGGCC repeat expansion in C9ORF72. Pathologically, FTD is characterised by abnormal protein accumulations in neurons and glia. These aggregates can be composed of the microtubule-associated protein tau (observed in FTD with MAPT mutations), the DNA/RNA-binding protein TDP-43 (seen in FTD with mutations in GRN or C9ORF72 repeat expansions) or dipeptide proteins generated by repeat associated non-ATG translation of the C9ORF72 repeat expansion. There are currently no disease-modifying therapies for FTD and the availability of in vitro models that recapitulate pathologies in a disease-relevant cell type would accelerate the development of novel therapeutics. It is now possible to generate patient-specific stem cells through the reprogramming of somatic cells from a patient with a genotype/phenotype of interest into induced pluripotent stem cells (iPSCs). iPSCs can subsequently be differentiated into a plethora of cell types including neurons, astrocytes and microglia. Using this approach has allowed researchers to generate in vitro models of genetic FTD in human cell types that are largely inaccessible during life. In this review we explore the recent progress in the use of iPSCs to model FTD, and consider the merits, limitations and future prospects of this approach.


Frontotemporal Dementia/genetics , Induced Pluripotent Stem Cells/metabolism , tau Proteins/genetics , Axons/metabolism , Biological Transport , C9orf72 Protein/genetics , C9orf72 Protein/physiology , Cell Differentiation , Cellular Reprogramming Techniques , DNA Repeat Expansion , DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Humans , Introns/genetics , Microtubules/physiology , Mitochondria/physiology , Models, Genetic , Mutation, Missense , Nerve Degeneration , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Organoids , Progranulins/genetics , Progranulins/physiology , Protein Aggregation, Pathological , Protein Isoforms , Protein Splicing , Reactive Oxygen Species , tau Proteins/chemistry , tau Proteins/metabolism
4.
Endocr J ; 67(8): 819-825, 2020 Aug 28.
Article En | MEDLINE | ID: mdl-32321883

Pregnant women with obesity are at increased risk of parturition dysfunction; however, the biological mechanism has remained unknown. We hypothesized that molecules circulating in the serum of pregnant women with obesity may induce the aberrant expression of contraction-associated proteins (CAPs), leading to insufficient uterine contractions. This study aimed to investigate the effects of maternal serum on CAPs expression by human uterine smooth muscle cells (UtSMCs) and elucidate the influence of maternal obesity. Blood samples were collected from singleton pregnant women at 36-41 weeks of gestation before the onset of labor. UtSMCs were incubated in the serum, and the mRNA expressions of PTGFR, OXTR, GJA1, and PTGS2 were examined by RT-PCR. Progranulin (PGRN) is a circulating glycoprotein associated with insulin resistance characterized by the accumulation of visceral fat. The serum PGRN levels of the samples were measured by ELISA. After incubated with PGRN (100-1,000 ng/mL), mRNA expression of PTGFR, OXTR, and GJA1 and protein expression of CX43 were examined by RT-PCR and western blotting, respectively. The mRNA expressions of PTGFR, OXTR, and GJA1 showed significantly negative correlations with gestational weight gain (GWG). Serum PGRN levels showed a significantly positive correlation with GWG. High levels of PGRN suppressed the mRNA expression of GJA1 and the protein expression of CX43. The change in maternal serum induced by GWG suppressed the CAPs expression by UtSMCs. PGRN is one of the factors in the serum responsible for inhibiting the expression of CX43.


Contractile Proteins/genetics , Gestational Weight Gain , Myocytes, Smooth Muscle/metabolism , Progranulins/physiology , Uterus/metabolism , Adult , Cells, Cultured , Contractile Proteins/metabolism , Culture Media, Conditioned/pharmacology , Female , Gene Expression/drug effects , Gestational Weight Gain/genetics , Gestational Weight Gain/physiology , Humans , Myocytes, Smooth Muscle/drug effects , Obesity/genetics , Obesity/metabolism , Obesity/physiopathology , Parturition/blood , Parturition/metabolism , Pregnancy , Pregnancy Complications/genetics , Pregnancy Complications/metabolism , Pregnancy Complications/physiopathology , Progranulins/blood , Progranulins/pharmacology , Serum/physiology , Uterine Contraction/genetics , Uterine Contraction/metabolism , Uterus/cytology
5.
J Mol Endocrinol ; 64(3): 181-193, 2020 04.
Article En | MEDLINE | ID: mdl-31990656

Progranulin (PGRN), a multifunctional protein implicated in embryonic development and immune response, was recently introduced as a novel marker of chronic inflammation related with insulin resistance in obesity and type 2 diabetes mellitus. However, the potential mechanisms of PGRN on insulin signaling pathways are poorly understood. In this study, PGRN mediated the chemotaxis of RAW264.7, impaired insulin action and stimulated production of inflammatory factors in adipocytes, which was accompanied by increased c-Jun N-terminal kinase (JNK) activation and serine phosphorylation of insulin receptor substrate-1. PGRN knockdown partially led to an increase in insulin action as well as a decrease in the JNK activation and extracellular signal-regulated kinase phosphorylation in cells exposed to tumor-necrosis factor-α (TNF-α). Meanwhile, PGRN treatment resulted in an elevation of transcription factor nuclear factor κB (NF-κB) nuclear translocation and acetylation, and increased Il-1b, Il6, Tnf-a expression, whereas NF-κB inhibition reversed PGRN-induced insulin action impairment and inflammatory gene expression. Finally, we showed that sirtuin 1 (SIRT1) expression was downregulated by PGRN treatment, whereas SIRT1 overexpression improved PGRN-induced insulin resistance, NF-κB activation, and inflammatory gene expression. Our results suggest that PGRN regulates adipose tissue inflammation possibly by controlling the gain of proinflammatory transcription in a SIRT1-NF-κB dependent manner in response to inducers such as fatty acids and endoplasmic reticulum stress.


Adipose Tissue/metabolism , Inflammation/genetics , Insulin Resistance/genetics , Progranulins/physiology , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Inflammation/metabolism , Inflammation/pathology , Insulin/metabolism , Insulin/pharmacology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , RAW 264.7 Cells , Signal Transduction/drug effects , Signal Transduction/genetics , Sirtuin 1/genetics , Sirtuin 1/metabolism
6.
Neurosci Lett ; 720: 134755, 2020 02 16.
Article En | MEDLINE | ID: mdl-31945450

Our previous investigations revealed that progranulin (PGRN) is a lysosomal protein involved in hippocampal neurogenesis and neuroinflammation. However, the possible involvement of PGRN in regulating inflammatory response and mediating neuronal activity is still not well-defined. Here, we demonstrate that PGRN deficiency enhances the age-dependent increase of neuronal activity in the paraventricular nucleus (PVN) of the hypothalamus. Aging increased neuronal activity in the PVN of the hypothalamus, and PGRN deficiency enhanced the effects of age on hypothalamic neuronal activity. Aging increased the lysosomal biogenesis and inflammatory response in microglia, which was also aggravated in PGRN-knockout mice. Moreover, PGRN deficiency enhanced interleukin-1 beta and lysosomal genes levels. These results suggest that PGRN deficiency may enhance the age-dependent increase of neuronal activity possibly because PGRN facilitates immunological responses through regulating lysosomal function.


Encephalitis/physiopathology , Neurons/physiology , Paraventricular Hypothalamic Nucleus/physiology , Progranulins/physiology , Animals , Cytokines/metabolism , Lysosomes/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Microglia/physiology , Progranulins/genetics
7.
PLoS Pathog ; 15(10): e1008062, 2019 10.
Article En | MEDLINE | ID: mdl-31585000

Type I interferons (IFNs) play a critical role in host defense against influenza virus infection, and the mechanism of influenza virus to evade type I IFNs responses remains to be fully understood. Here, we found that progranulin (PGRN) was significantly increased both in vitro and in vivo during influenza virus infection. Using a PGRN knockdown assay and PGRN-deficient mice model, we demonstrated that influenza virus-inducing PGRN negatively regulated type I IFNs production by inhibiting the activation of NF-κB and IRF3 signaling. Furthermore, we showed that PGRN directly interacted with NF-κB essential modulator (NEMO) via its Grn CDE domains. We also verified that PGRN recruited A20 to deubiquitinate K63-linked polyubiquitin chains on NEMO at K264. In addition, we found that macrophage played a major source of PGRN during influenza virus infection, and PGRN neutralizing antibodies could protect against influenza virus-induced lethality in mice. Our data identify a PGRN-mediated IFN evasion pathway exploited by influenza virus with implication in antiviral applications. These findings also provide insights into the functions and crosstalk of PGRN in innate immunity.


Antiviral Agents/pharmacology , Immune Evasion/immunology , Immunity, Innate/immunology , Interferon Type I/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Progranulins/physiology , Animals , Cells, Cultured , Down-Regulation , Host-Pathogen Interactions , Male , Mice , Mice, Knockout , NF-kappa B , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Signal Transduction
8.
Endocrinology ; 160(9): 2024-2037, 2019 09 01.
Article En | MEDLINE | ID: mdl-31237618

Progranulin (PGRN) is best known as a glial protein for which deficiency leads to the most common inherited form of frontotemporal dementia. Recently, PGRN has been found to be an adipokine associated with diet-induced obesity and insulin resistance. Therefore, PGRN may have homeostatic effects on bone because PGRN is reported to promote the differentiation of bone-resorbing osteoclasts. We investigated the actions of PGRN on bone using PGRN gene (Grn) knockout (KO) mice and transgenic mice with PGRN mutation and surprisingly found that loss of PGRN prevented the bone loss in female mice induced by aging and estrogen deficiency, whereas it had no effect on male bones during aging. Strikingly, bone formation was increased in female (but not male) PGRN KO mice. We also found that loss of PGRN inhibited bone resorption and osteoclastogenesis in both male and female mice and promoted the production of osteogenic factors in osteoclast lineage cells. These results indicate that PGRN serves to uncouple bone turnover in female mice by promoting bone resorption and suppressing bone formation. Furthermore, we demonstrated that microglial cells/macrophages, but not adipocytes, are an important source of PGRN in producing negative skeletal effects in females. Targeting PGRN production by microglial cells/macrophage-lineage cells may provide a therapeutic approach for the treatment of osteoporosis in females.


Osteogenesis/physiology , Progranulins/physiology , Animals , Bone Resorption/etiology , Estrogens/deficiency , Female , Male , Mice , Mice, Inbred C57BL , Progranulins/genetics , Sex Factors
9.
J Cosmet Dermatol ; 18(6): 2019-2026, 2019 Dec.
Article En | MEDLINE | ID: mdl-31091001

BACKGROUND/OBJECTIVES: Progranulin (PGRN) is a secreted glycoprotein that was investigated in many skin diseases. It plays an important role in inflammatory response and autophagy which could be mediated through Wnt/ß-catenin pathway. However, the role of PGRN in pathogenesis of psoriasis has not been clearly well-known. Therefore, we aimed by this study to investigate the possible role of progranulin in psoriasis pathogenesis through evaluation of its immunohistochemical expression in lesional and perilesional skin of psoriasis patients and to investigate if its hypothesized role is mediated through ß-catenin or not. METHODS: This case-control study was carried out on 37 patients presented with variable degrees of psoriasis vulgaris severity vs 37 age and sex-matched apparently healthy volunteers. Psoriasis area and severity index (PASI) score was used to evaluate the severity of psoriasis. From all cases (lesional and perilesional) and controls, skin biopsies were taken for histopathological and immunohistochemical evaluation of PGRN and ß-catenin. RESULTS: There was a significant stepwise upregulation of PGRN from controls (76.2 ± 11.9) to perilesional (178.7 ± 11.8) and lesional (242.7 ± 12.7) psoriatic skin (P < 0.001). PGRN expression was significantly correlated with psoriasis severity (r = 0.61; P < 0.001). ß-catenin showed a significant stepwise downregulation from control (210.0 ± 19.3) to perilesional (131.4 ± 9.2) and lesional (97.3 ± 11.5) psoriatic skin(P < 0.001). There was a significant negative correlation between PGRN and ß-catenin expression in psoriatic skin (P < 0.001). CONCLUSIONS: Progranulin has a pro-inflammatory effect in the psoriasis pathogenesis, which could be mediated through a decreasing ß-catenin expression in psoriasis. PGRN may be used as a target for immunotherapy in psoriasis management program.


Progranulins/physiology , Psoriasis/etiology , beta Catenin/physiology , Adult , Case-Control Studies , Female , Humans , Immunohistochemistry , Male , Middle Aged , Progranulins/analysis , Psoriasis/metabolism , Severity of Illness Index , Skin/chemistry , Young Adult , beta Catenin/analysis
10.
Cytokine Growth Factor Rev ; 45: 53-64, 2019 02.
Article En | MEDLINE | ID: mdl-30733059

Progranulin (PGRN), a widely expressed glycoprotein with pleiotropic function, has been linked to a host of physiological processes and diverse pathological states. A series of contemporary preclinical disease models and clinical trials have evaluated various therapeutic strategies targeting PGRN, highlighting PGRN as a promising therapeutic target. Herein we summarize available knowledge of PGRN targeting in various kinds of diseases, including common neurological diseases, inflammatory autoimmune diseases, cancer, tissue repair, and rare lysosomal storage diseases, with a focus on the functional domain-oriented drug development strategies. In particular, we emphasize the role of extracellular PGRN as a non-conventional, extracellular matrix bound, growth factor-like conductor orchestrating multiple membrane receptors and intracellular PGRN as a chaperone/co-chaperone that mediates the folding and traffic of its various binding partners.


Autoimmune Diseases/drug therapy , Lysosomal Storage Diseases/drug therapy , Neoplasms/drug therapy , Nervous System Diseases/drug therapy , Progranulins/physiology , Animals , Clinical Trials as Topic , Disease Models, Animal , Drug Development , Humans , Inflammation/drug therapy , Lysosomes/enzymology , Mice , Molecular Chaperones/physiology , Molecular Targeted Therapy , Neurodegenerative Diseases/drug therapy
11.
Acta Neuropathol ; 137(6): 879-899, 2019 06.
Article En | MEDLINE | ID: mdl-30739198

Frontotemporal lobar degeneration with neuronal inclusions of the TAR DNA-binding protein 43 (FTLD-TDP) represents the most common pathological subtype of FTLD. We established the international FTLD-TDP whole-genome sequencing consortium to thoroughly characterize the known genetic causes of FTLD-TDP and identify novel genetic risk factors. Through the study of 1131 unrelated Caucasian patients, we estimated that C9orf72 repeat expansions and GRN loss-of-function mutations account for 25.5% and 13.9% of FTLD-TDP patients, respectively. Mutations in TBK1 (1.5%) and other known FTLD genes (1.4%) were rare, and the disease in 57.7% of FTLD-TDP patients was unexplained by the known FTLD genes. To unravel the contribution of common genetic factors to the FTLD-TDP etiology in these patients, we conducted a two-stage association study comprising the analysis of whole-genome sequencing data from 517 FTLD-TDP patients and 838 controls, followed by targeted genotyping of the most associated genomic loci in 119 additional FTLD-TDP patients and 1653 controls. We identified three genome-wide significant FTLD-TDP risk loci: one new locus at chromosome 7q36 within the DPP6 gene led by rs118113626 (p value = 4.82e - 08, OR = 2.12), and two known loci: UNC13A, led by rs1297319 (p value = 1.27e - 08, OR = 1.50) and HLA-DQA2 led by rs17219281 (p value = 3.22e - 08, OR = 1.98). While HLA represents a locus previously implicated in clinical FTLD and related neurodegenerative disorders, the association signal in our study is independent from previously reported associations. Through inspection of our whole-genome sequence data for genes with an excess of rare loss-of-function variants in FTLD-TDP patients (n ≥ 3) as compared to controls (n = 0), we further discovered a possible role for genes functioning within the TBK1-related immune pathway (e.g., DHX58, TRIM21, IRF7) in the genetic etiology of FTLD-TDP. Together, our study based on the largest cohort of unrelated FTLD-TDP patients assembled to date provides a comprehensive view of the genetic landscape of FTLD-TDP, nominates novel FTLD-TDP risk loci, and strongly implicates the immune pathway in FTLD-TDP pathogenesis.


Nerve Tissue Proteins/genetics , TDP-43 Proteinopathies/genetics , Aged , DNA Repeat Expansion , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Female , Frontal Lobe/metabolism , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/immunology , Genetic Predisposition to Disease , Genome-Wide Association Study , HLA-DQ Antigens/genetics , Humans , Intracellular Signaling Peptides and Proteins , Loss of Function Mutation , Male , Middle Aged , Nerve Tissue Proteins/physiology , Potassium Channels/genetics , Progranulins/genetics , Progranulins/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Proteins/genetics , Proteins/physiology , RNA, Messenger/biosynthesis , Risk Factors , Sequence Analysis, RNA , Societies, Scientific , TDP-43 Proteinopathies/immunology , White People/genetics
12.
Neuron ; 97(4): 796-805.e5, 2018 02 21.
Article En | MEDLINE | ID: mdl-29398357

Elimination of redundant synapses formed early in development and strengthening of necessary connections are crucial for shaping functional neural circuits. Purkinje cells (PCs) in the neonatal cerebellum are innervated by multiple climbing fibers (CFs) with similar strengths. A single CF is strengthened whereas the other CFs are eliminated in each PC during postnatal development. The underlying mechanisms, particularly for the strengthening of single CFs, are poorly understood. Here we report that progranulin, a multi-functional growth factor implicated in the pathogenesis of frontotemporal dementia, strengthens developing CF synaptic inputs and counteracts their elimination from postnatal day 11 to 16. Progranulin derived from PCs acts retrogradely onto its putative receptor Sort1 on CFs. This effect is independent of semaphorin 3A, another retrograde signaling molecule that counteracts CF synapse elimination. We propose that progranulin-Sort1 signaling strengthens and maintains developing CF inputs, and may contribute to selection of single "winner" CFs that survive synapse elimination.


Adaptor Proteins, Vesicular Transport/physiology , Cerebellum/growth & development , Dendrites/physiology , Intercellular Signaling Peptides and Proteins/physiology , Neuronal Plasticity , Purkinje Cells/physiology , Synapses/physiology , Animals , Excitatory Postsynaptic Potentials , Female , Granulins , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Progranulins/physiology , Rats, Sprague-Dawley , Semaphorin-3A/physiology , Signal Transduction
13.
Cell Syst ; 4(4): 404-415.e5, 2017 04 26.
Article En | MEDLINE | ID: mdl-28330615

Human age-associated traits, such as cognitive decline, can be highly variable across the population, with some individuals exhibiting traits that are not expected at a given chronological age. Here we present differential aging (Δ-aging), an unbiased method that quantifies individual variability in age-associated phenotypes within a tissue of interest, and apply this approach to the analysis of existing transcriptome-wide cerebral cortex gene expression data from several cohorts totaling 1,904 autopsied human brain samples. We subsequently performed a genome-wide association study and identified the TMEM106B and GRN gene loci, previously associated with frontotemporal dementia, as determinants of Δ-aging in the cerebral cortex with genome-wide significance. TMEM106B risk variants are associated with inflammation, neuronal loss, and cognitive deficits, even in the absence of known brain disease, and their impact is highly selective for the frontal cerebral cortex of older individuals (>65 years). The methodological framework we describe can be broadly applied to the analysis of quantitative traits associated with aging or with other parameters.


Aging/genetics , Cerebral Cortex/metabolism , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Progranulins/genetics , Age Factors , Aged , Aged, 80 and over , Aging/pathology , Aging/physiology , Cerebral Cortex/pathology , Cohort Studies , Gene Expression Profiling , Genome-Wide Association Study , Humans , Immunity, Innate/genetics , Membrane Proteins/metabolism , Membrane Proteins/physiology , Nerve Degeneration/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Phenotype , Progranulins/metabolism , Progranulins/physiology
...